1
|
McEwen DP, Ray P, Nancarrow DJ, Wang Z, Kasturirangan S, Abdullah S, Balan A, Hoskeri R, Thomas D, Lawrence TS, Beer DG, Lagisetty KH, Ray D. ISG15/GRAIL1/CD3 axis influences survival of patients with esophageal adenocarcinoma. JCI Insight 2024; 9:e179315. [PMID: 38781019 PMCID: PMC11383178 DOI: 10.1172/jci.insight.179315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024] Open
Abstract
Immunosuppression is a common feature of esophageal adenocarcinoma (EAC) and has been linked to poor overall survival (OS). We hypothesized that upstream factors might negatively influence CD3 levels and T cell activity, thus promoting immunosuppression and worse survival. We used clinical data and patient samples of those who progressed from Barrett's to dysplasia to EAC, investigated gene (RNA-Seq) and protein (tissue microarray) expression, and performed cell biology studies to delineate a pathway impacting CD3 protein stability that might influence EAC outcome. We showed that the loss of both CD3-ε expression and CD3+ T cell number correlated with worse OS in EAC. The gene related to anergy in lymphocytes isoform 1 (GRAIL1), which is the prominent isoform in EACs, degraded (ε, γ, δ) CD3s and inactivated T cells. In contrast, isoform 2 (GRAIL2), which is reduced in EACs, stabilized CD3s. Further, GRAIL1-mediated CD3 degradation was facilitated by interferon-stimulated gene 15 (ISG15), a ubiquitin-like protein. Consequently, the overexpression of a ligase-dead GRAIL1, ISG15 knockdown, or the overexpression of a conjugation-defective ISG15-leucine-arginine-glycine-glycine mutant could increase CD3 levels. Together, we identified an ISG15/GRAIL1/mutant p53 amplification loop negatively influencing CD3 levels and T cell activity, thus promoting immunosuppression in EAC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dafydd Thomas
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - David G Beer
- Department of Surgery, Section of Thoracic Surgery
| | | | | |
Collapse
|
2
|
Lu H, Cao LL, Ballout F, Belkhiri A, Peng D, Chen L, Chen Z, Soutto M, Wang TC, Que J, Giordano S, Washington MK, Chen S, McDonald OG, Zaika A, El-Rifai W. Reflux conditions induce E-cadherin cleavage and EMT via APE1 redox function in oesophageal adenocarcinoma. Gut 2023; 73:47-62. [PMID: 37734913 PMCID: PMC10872865 DOI: 10.1136/gutjnl-2023-329455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023]
Abstract
OBJECTIVE Chronic gastro-oesophageal reflux disease, where acidic bile salts (ABS) reflux into the oesophagus, is the leading risk factor for oesophageal adenocarcinoma (EAC). We investigated the role of ABS in promoting epithelial-mesenchymal transition (EMT) in EAC. DESIGN RNA sequencing data and public databases were analysed for the EMT pathway enrichment and patients' relapse-free survival. Cell models, pL2-IL1β transgenic mice, deidentified EAC patients' derived xenografts (PDXs) and tissues were used to investigate EMT in EAC. RESULTS Analysis of public databases and RNA-sequencing data demonstrated significant enrichment and activation of EMT signalling in EAC. ABS induced multiple characteristics of the EMT process, such as downregulation of E-cadherin, upregulation of vimentin and activation of ß-catenin signalling and EMT-transcription factors. These were associated with morphological changes and enhancement of cell migration and invasion capabilities. Mechanistically, ABS induced E-cadherin cleavage via an MMP14-dependent proteolytic cascade. Apurinic/apyrimidinic endonuclease (APE1), also known as redox factor 1, is an essential multifunctional protein. APE1 silencing, or its redox-specific inhibitor (E3330), downregulated MMP14 and abrogated the ABS-induced EMT. APE1 and MMP14 coexpression levels were inversely correlated with E-cadherin expression in human EAC tissues and the squamocolumnar junctions of the L2-IL1ß transgenic mouse model of EAC. EAC patients with APE1high and EMThigh signatures had worse relapse-free survival than those with low levels. In addition, treatment of PDXs with E3330 restrained EMT characteristics and suppressed tumour invasion. CONCLUSION Reflux conditions promote EMT via APE1 redox-dependent E-cadherin cleavage. APE1-redox function inhibitors can have a therapeutic role in EAC.
Collapse
Affiliation(s)
- Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Long Long Cao
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Farah Ballout
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - DunFa Peng
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Lei Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Zheng Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mohammed Soutto
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Veterans Affairs, VA Miami Healthcare System, Miami, FL, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, NY, USA
| | - Silvia Giordano
- Department of Oncology, University of Torino and Candiolo Cancer Institute, Candiolo, Italy
| | - Mary Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Steven Chen
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Oliver Gene McDonald
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Veterans Affairs, VA Miami Healthcare System, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Veterans Affairs, VA Miami Healthcare System, Miami, FL, USA
| |
Collapse
|
3
|
Yi Q, Zhao Y, Xia R, Wei Q, Chao F, Zhang R, Bian P, Lv L. TRIM29 hypermethylation drives esophageal cancer progression via suppression of ZNF750. Cell Death Discov 2023; 9:191. [PMID: 37365152 DOI: 10.1038/s41420-023-01491-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Esophageal cancer (ESCA) is the seventh most frequent and deadly neoplasm. Due to the lack of early diagnosis and high invasion/metastasis, the prognosis of ESCA remains very poor. Herein, we identify skin-related signatures as the most deficient signatures in invasive ESCA, which are regulated by the transcription factor ZNF750. Of note, we find that TRIM29 level strongly correlated with the expression of many genes in the skin-related signatures, including ZNF750. TRIM29 is significantly down-regulated due to hypermethylation of its promoter in both ESCA and precancerous lesions compared to normal tissues. Low TRIM29 expression and high methylation levels of its promoter are associated with malignant progression and poor clinical outcomes in ESCA patients. Functionally, TRIM29 overexpression markedly hinders proliferation, migration, invasion, and epithelial-mesenchymal transition of esophageal cancer cells, whereas opposing results are observed when TRIM29 is silenced in vitro. In addition, TRIM29 inhibits metastasis in vivo. Mechanistically, TRIM29 downregulation suppresses the expression of the tumor suppressor ZNF750 by activating the STAT3 signaling pathway. Overall, our study demonstrates that TRIM29 expression and its promoter methylation status could be potential early diagnostic and prognostic markers. It highlights the role of the TRIM29-ZNF750 signaling axis in modulating tumorigenesis and metastasis of esophageal cancer.
Collapse
Affiliation(s)
- Qiyi Yi
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
| | - Yujia Zhao
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
- Department of education training, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ran Xia
- Department of Cancer Epigenetics Program, Anhui Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230031, Hefei, Anhui, China
| | - Qinqin Wei
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
| | - Fengmei Chao
- Department of Cancer Epigenetics Program, Anhui Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230031, Hefei, Anhui, China
| | - Rui Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui University of Chinese Medicine, 230031, Hefei, Anhui, China
| | - Po Bian
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China.
| | - Lei Lv
- Department of Cancer Epigenetics Program, Anhui Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230031, Hefei, Anhui, China.
| |
Collapse
|
4
|
Schleser SW, Ghosh H, Hörner G, Seib J, Bhattacharyya S, Weber B, Schobert R, Dandawate P, Biersack B. New 4,5-Diarylimidazol-2-ylidene-iodidogold(I) Complexes with High Activity against Esophageal Adenocarcinoma Cells. Int J Mol Sci 2023; 24:5738. [PMID: 36982817 PMCID: PMC10052191 DOI: 10.3390/ijms24065738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Inspired by the vascular-disrupting agent combretastatin A-4 and recently published anticancer active N-heterocyclic carbene (NHC) complexes of Au(I), a series of new iodidogold(I)-NHC complexes was synthesized and characterized. The iodidogold(I) complexes were synthesized by a route involving van Leusen imidazole formation and N-alkylation, followed by complexation with Ag2O, transmetalation with chloro(dimethylsulfide)gold(I) [Au(DMS)Cl], and anion exchange with KI. The target complexes were characterized by IR spectroscopy, 1H and 13C NMR spectroscopy, and mass spectrometry. The structure of 6c was validated via single-crystal X-ray diffraction. A preliminary anticancer screening of the complexes using two esophageal adenocarcinoma cell lines showed promising nanomolar activities for certain iodidogold(I) complexes accompanied with apoptosis induction, as well as c-Myc and cyclin D1 suppression in esophageal adenocarcinoma cells treated with the most promising derivative 6b.
Collapse
Affiliation(s)
- Sebastian W. Schleser
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Hindole Ghosh
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Gerald Hörner
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Jonathan Seib
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Sangita Bhattacharyya
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Birgit Weber
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Prasad Dandawate
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
5
|
Zhang Y, He Q. The role of SELENBP1 and its epigenetic regulation in carcinogenic progression. Front Genet 2022; 13:1027726. [PMID: 36386843 PMCID: PMC9663989 DOI: 10.3389/fgene.2022.1027726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
The initiation and progression of cancer is modulated through diverse genetic and epigenetic modifications. The epigenetic machinery regulates gene expression through intertwined DNA methylation, histone modifications, and miRNAs without affecting their genome sequences. SELENBP1 belongs to selenium-binding proteins and functions as a tumor suppressor. Its expression is significantly downregulated and correlates with carcinogenic progression and poor survival in various cancers. The role of SELENBP1 in carcinogenesis has not been fully elucidated, and its epigenetic regulation remains poorly understood. In this review, we summarize recent findings on the function and regulatory mechanisms of SELENBP1 during carcinogenic progression, with an emphasis on epigenetic mechanisms. We also discuss the potential cancer treatment targeting epigenetic modification of SELENBP1, either alone or in combination with selenium-containing compounds or dietary selenium.
Collapse
|
6
|
Wang L, Liu Z, Liang R, Wang W, Zhu R, Li J, Xing Z, Weng S, Han X, Sun YL. Comprehensive machine-learning survival framework develops a consensus model in large-scale multicenter cohorts for pancreatic cancer. eLife 2022; 11:e80150. [PMID: 36282174 PMCID: PMC9596158 DOI: 10.7554/elife.80150] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/15/2022] [Indexed: 11/13/2022] Open
Abstract
As the most aggressive tumor, the outcome of pancreatic cancer (PACA) has not improved observably over the last decade. Anatomy-based TNM staging does not exactly identify treatment-sensitive patients, and an ideal biomarker is urgently needed for precision medicine. Based on expression files of 1280 patients from 10 multicenter cohorts, we screened 32 consensus prognostic genes. Ten machine-learning algorithms were transformed into 76 combinations, of which we selected the optimal algorithm to construct an artificial intelligence-derived prognostic signature (AIDPS) according to the average C-index in the nine testing cohorts. The results of the training cohort, nine testing cohorts, Meta-Cohort, and three external validation cohorts (290 patients) consistently indicated that AIDPS could accurately predict the prognosis of PACA. After incorporating several vital clinicopathological features and 86 published signatures, AIDPS exhibited robust and dramatically superior predictive capability. Moreover, in other prevalent digestive system tumors, the nine-gene AIDPS could still accurately stratify the prognosis. Of note, our AIDPS had important clinical implications for PACA, and patients with low AIDPS owned a dismal prognosis, higher genomic alterations, and denser immune cell infiltrates as well as were more sensitive to immunotherapy. Meanwhile, the high AIDPS group possessed observably prolonged survival, and panobinostat may be a potential agent for patients with high AIDPS. Overall, our study provides an attractive tool to further guide the clinical management and individualized treatment of PACA.
Collapse
Affiliation(s)
- Libo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ruopeng Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Weijie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Rongtao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Jian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu-ling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| |
Collapse
|
7
|
Zhang X, Hong R, Bei L, Hu Z, Yang X, Song T, Chen L, Meng H, Niu G, Ke C. SELENBP1 inhibits progression of colorectal cancer by suppressing epithelial–mesenchymal transition. Open Med (Wars) 2022; 17:1390-1404. [PMID: 36117772 PMCID: PMC9438969 DOI: 10.1515/med-2022-0532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/19/2022] [Accepted: 07/11/2022] [Indexed: 01/13/2023] Open
Abstract
Selenium-binding protein 1 (SELENBP1) is frequently dysregulated in various malignancies including colorectal cancer (CRC); however, its roles in progression of CRCs and the underlying mechanism remain to be elucidated. In this study, we compared the expression of SELENBP1 between CRCs and colorectal normal tissues (NTs), as well as between primary and metastatic CRCs; we determined the association between SELENBP1 expression and CRC patient prognoses; we conducted both in vitro and in vivo experiments to explore the functional roles of SELENBP1 in CRC progression; and we characterized the potential underlying mechanisms associated with SELENBP1 activities. We found that the expression of SELENBP1 was significantly and consistently decreased in CRCs than that in adjacent NTs, while significantly and frequently decreased in metastatic than primary CRCs. High expression of SELENBP1 was an independent predictor of favorable prognoses in CRC patients. Overexpression of SELENBP1 suppressed, while silencing of SELENBP1 promoted cell proliferation, migration and invasion, and in vivo tumorigenesis of CRC. Mechanically, SELENBP1 may suppress CRC progression by inhibiting the epithelial–mesenchymal transition.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Runqi Hong
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Lanxin Bei
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhiqing Hu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Ximin Yang
- Department of Radiology, Dongying New District Hospital, Dongying, Shandong Province, 257000, P.R. China
| | - Tao Song
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Liang Chen
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - He Meng
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Gengming Niu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Chongwei Ke
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| |
Collapse
|
8
|
Zhao X, Gabriëls RY, Hooghiemstra WTR, Koller M, Meersma GJ, Buist-Homan M, Visser L, Robinson DJ, Tenditnaya A, Gorpas D, Ntziachristos V, Karrenbeld A, Kats-Ugurlu G, Fehrmann RSN, Nagengast WB. Validation of Novel Molecular Imaging Targets Identified by Functional Genomic mRNA Profiling to Detect Dysplasia in Barrett's Esophagus. Cancers (Basel) 2022; 14:cancers14102462. [PMID: 35626066 PMCID: PMC9139936 DOI: 10.3390/cancers14102462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Barrett’s esophagus (BE) is the precursor of esophageal adenocarcinoma (EAC). Dysplastic BE (DBE) has a higher progression risk to EAC compared to non-dysplastic BE (NDBE). However, the miss rates for the endoscopic detection of DBE remain high. Fluorescence molecular endoscopy (FME) can detect DBE and mucosal EAC by highlighting the tumor-specific expression of proteins. This study aimed to identify target proteins suitable for FME. Publicly available RNA expression profiles of EAC and NDBE were corrected by functional genomic mRNA (FGmRNA) profiling. Following a class comparison between FGmRNA profiles of EAC and NDBE, predicted, significantly upregulated genes in EAC were prioritized by a literature search. Protein expression of prioritized genes was validated by immunohistochemistry (IHC) on DBE and NDBE tissues. Near-infrared fluorescent tracers targeting the proteins were developed and evaluated ex vivo on fresh human specimens. In total, 1976 overexpressed genes were identified in EAC (n = 64) compared to NDBE (n = 66) at RNA level. Prioritization and IHC validation revealed SPARC, SULF1, PKCι, and DDR1 (all p < 0.0001) as the most attractive imaging protein targets for DBE detection. Newly developed tracers SULF1-800CW and SPARC-800CW both showed higher fluorescence intensity in DBE tissue compared to paired non-dysplastic tissue. This study identified SPARC, SULF1, PKCι, and DDR1 as promising targets for FME to differentiate DBE from NDBE tissue, for which SULF1-800CW and SPARC-800CW were successfully ex vivo evaluated. Clinical studies should further validate these findings.
Collapse
Affiliation(s)
- Xiaojuan Zhao
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
- Cancer Research Center Groningen, Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Ruben Y. Gabriëls
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
| | - Wouter T. R. Hooghiemstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Marjory Koller
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Gert Jan Meersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
- Cancer Research Center Groningen, Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Lydia Visser
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (L.V.); (A.K.); (G.K.-U.)
| | - Dominic J. Robinson
- Center for Optic Diagnostics and Therapy, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Anna Tenditnaya
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 80333 Munich, Germany; (A.T.); (D.G.); (V.N.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), 85764 Neuherberg, Germany
| | - Dimitris Gorpas
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 80333 Munich, Germany; (A.T.); (D.G.); (V.N.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), 85764 Neuherberg, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 80333 Munich, Germany; (A.T.); (D.G.); (V.N.)
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), 85764 Neuherberg, Germany
| | - Arend Karrenbeld
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (L.V.); (A.K.); (G.K.-U.)
| | - Gursah Kats-Ugurlu
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (L.V.); (A.K.); (G.K.-U.)
| | - Rudolf S. N. Fehrmann
- Cancer Research Center Groningen, Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Wouter B. Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (X.Z.); (R.Y.G.); (W.T.R.H.); (G.J.M.); (M.B.-H.)
- Correspondence: ; Tel.: +31-(50)-361-6161
| |
Collapse
|
9
|
Ming R, Li X, Wang E, Wei J, Liu B, Zhou P, Yu W, Zong S, Xiao H. The Prognostic Signature of Head and Neck Squamous Cell Carcinoma Constructed by Immune-Related RNA-Binding Proteins. Front Oncol 2022; 12:795781. [PMID: 35449571 PMCID: PMC9016149 DOI: 10.3389/fonc.2022.795781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to construct a prognostic signature consisting of immune-related RNA-binding proteins (RBPs) to predict the prognosis of patients with head and neck squamous cell carcinoma (HNSCC) effectively. Methods The transcriptome and clinical data of HNSCC were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. First, we ascertained the immunological differences in HNSCC, through single-sample gene set enrichment analysis, stromal and immune cells in malignant tumor tissues using expression data (ESTIMATE), and cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) deconvolution algorithm. Then we used univariate proportional hazards (Cox) regression analysis and least absolute shrinkage and selection operator (LASSO) Cox regression analysis to screen immune-related RBPs and acquire the risk score of each sample. Subsequently, we further investigated the difference in prognosis, immune status, and tumor mutation burden in high- and low-risk groups. Finally, the efficacy of immunotherapy was measured by the tumor immune dysfunction and exclusion (TIDE) score. Results We derived 15 immune-related RBPs, including FRMD4A, ASNS, RAB11FIP1, FAM120C, CFLAR, CTTN, PLEKHO1, SELENBP1, CHCHD2, NPM3, ATP2A3, CFDP1, IGF2BP2, NQO1, and DENND2D. There were significant differences in the prognoses of patients in the high- and low-risk groups in the training set (p < 0.001) and the validation set (p < 0.01). Furthermore, there were statistical differences between the high-risk group and low-risk group in immune cell infiltration and pathway and tumor mutation load (p < 0.001). In the end, we found that patients in the low-risk group were more sensitive to immunotherapy (p < 0.001), and then we screened 14 small-molecule chemotherapeutics with higher sensitivity to the high-risk group (p < 0.001). Conclusion The study constructed a prognostic signature of HNSCC, which might guide clinical immunotherapy in the future.
Collapse
Affiliation(s)
- Ruijie Ming
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangrui Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Enhao Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Wei
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenting Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shimin Zong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjun Xiao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Zhang X, Hong R, Bei L, Yang J, Zhao X, Hu Z, Chen L, Meng H, Zhang Q, Niu G, Yue Y, Ke C. Selenium binding protein 1 inhibits tumor angiogenesis in colorectal cancers by blocking the Delta-like ligand 4/Notch1 signaling pathway. Transl Oncol 2022; 18:101365. [PMID: 35158204 PMCID: PMC8850798 DOI: 10.1016/j.tranon.2022.101365] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 01/03/2023] Open
Abstract
SELENBP1 localizes to vessels and is suppressed in tumor vessels. SELENBP1 inhibits in vivo and in vitro angiogenesis. SELENBP1 antagonizes tumor angiogenesis by blocking the DLL4/Notch1 signaling pathway. SELENBP1 is a candidate target to treat bevacizumab-resistance in colorectal cancer.
Background Selenium binding protein 1 (SELENBP1) is frequently downregulated in malignancies such as colorectal cancer (CRC), however, whether it is involved in tumor angiogenesis is still unknown. Methods We analyzed the expression and localization of SELENBP1 in vessels from CRC and neighboring tissues. We investigated the in vitro and in vivo activity of SELENBP1 in angiogenesis and explored the underlying mechanism. Results SELENBP1 was localized to endothelial cells in addition to glandular cells, while its vascular expression was decreased in tumor vessels compared to that in vessels from neighboring non-tumor tissues. Gain-of-function and loss-of-function experiments demonstrated that SELENBP1 inhibited angiogenesis in vitro, and blocked communications between HUVECs and CRC cells. Overexpression of SELENBP1 in CRC cells inhibited tumor growth and angiogenesis, and enhanced bevacizumab-sensitivity in a mouse subcutaneous xenograft model. Mechanic analyses revealed that SELENBP1 may suppress tumor angiogenesis by binding with Delta-like ligand 4 (DLL4) and antagonizing the DLL4/Notch1 signaling pathway. The inhibitory effects of SELENBP1 on in vitro angiogenesis could largely be rescued by DLL4. Conclusion These results revealed a novel role of SELENBP1 as a potential tumor suppressor that antagonizes tumor angiogenesis in CRC by intervening the DLL4/Notch1 signaling pathway.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - Runqi Hong
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - Lanxin Bei
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ju Yang
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiaomei Zhao
- Department of Medicine, Dongying New District Hospital, Dongying, Shandong 257000, China
| | - Zhiqing Hu
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - Liang Chen
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China
| | - He Meng
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Zhang
- Department of Orthopedics, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221300, China
| | - Gengming Niu
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China.
| | - Ying Yue
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China.
| | - Chongwei Ke
- Department of General Surgery, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai 200240, China.
| |
Collapse
|
11
|
Panda A, Bhanot G, Ganesan S, Bajpai M. Gene Expression in Barrett's Esophagus Cell Lines Resemble Esophageal Squamous Cell Carcinoma Instead of Esophageal Adenocarcinoma. Cancers (Basel) 2021; 13:5971. [PMID: 34885081 PMCID: PMC8656995 DOI: 10.3390/cancers13235971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is strongly associated with Barrett's esophagus (BE), a pre-malignant condition resulting from gastric reflux. Esophageal squamous cell carcinoma (ESCC), the other major subtype of esophageal cancer, shows strong association with smoking and alcohol intake and no association with gastric reflux. In this study, we constructed and validated gene expression signatures of EAC vs. ESCC tumors using publicly available datasets, and subsequently assessed the enrichment levels of these signatures in commonly used EAC and ESCC cell lines, normal esophageal tissues and normal esophageal cell lines, and primary BE tissues and BE cell lines. We found that unlike ESCC cell lines which were quite similar to primary ESCC tumors, EAC cell lines were considerably different from primary EAC tumors but still more similar to EAC tumors than ESCC tumors, as the genes up in EAC vs. ESCC (EAChi) had considerably lower expression in EAC cell lines than EAC tumors. However, more surprisingly, unlike various normal cell lines (EPC2, Het-1A) which were very similar to various tissues from normal esophagus, BE cell lines (BAR-T, CP-A) were extremely different from primary BE tissues, as BE cell lines had substantially lower levels of EAChi and substantially higher levels of ESCChi gene expression. This ESCC-like profile of the BAR-T remained unaltered even after prolonged exposure to an acidic bile mixture in vitro resulting in malignant transformation of this cell line. However, primary BE tissues had EAC-like gene expression profiles as expected. Only one EAC case from the Cancer Genome Atlas resembled BE cell lines, and while it had the clinical profile and some mutational features of EAC, it had some mutational features, the copy number alteration profile, and the gene expression profile of ESCC instead. These incomprehensible changes in gene expression patterns may result in ambiguous changes in the phenotype and warrants careful evaluation to inform selection of appropriate in vitro tools for future studies on esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Anshuman Panda
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (A.P.); (G.B.); (S.G.)
| | - Gyan Bhanot
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
- Department of Physics and Astronomy, Rutgers University, Piscataway, NJ 08854, USA
| | - Shridar Ganesan
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (A.P.); (G.B.); (S.G.)
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine—Medical Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Manisha Bajpai
- Department of Medicine—Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Cancer Pharmacology Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
12
|
Zeng H, Zhao X, Tang C. Downregulation of SELENBP1 enhances oral squamous cell carcinoma chemoresistance through KEAP1-NRF2 signaling. Cancer Chemother Pharmacol 2021; 88:223-233. [PMID: 33907880 DOI: 10.1007/s00280-021-04284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/17/2021] [Indexed: 09/29/2022]
Abstract
PURPOSE Limited value is achieved in systemic chemotherapy for oral squamous cell carcinoma (OSCC), due to cancer cell resistance against cytotoxic agents. Tumor suppressor activities of selenium-binding protein 1 (SELENBP1) have been shown in multiple human cancers except for OSCC. The aim of this study is to clarify the biological functions and potential mechanism of SELENBP1 in OSCC. METHODS SELENBP1 expression and its clinical significance in OSCC were analyzed from The Cancer Genome Atlas (TCGA) database. Quantitative polymerase chain reaction (qPCR) or western blot was applied to determine SELENBP1, NRF2 and KEAP1 mRNA or protein levels. Sulforhodamine B assay (SRB) was performed to examine the cytotoxic effects of 5-fluorouracil (5-FU) and cisplatin on OSCC cells. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to investigate the role of SELENBP1 in KEAP1 transcription. RESULTS SELENBP1 downregulation is positively correlated with a poor prognosis for OSCC patients. SELENBP1 knockdown enhances resistance of OSCC cells to 5-FU and cisplatin, while SENENBP1 overexpression displays the opposite effects. Mechanistically, SELENBP1 reduces NRF2 protein levels by promoting its polyubiquitination and degradation. SELENBP1 induces KEAP1 transcription by binding to KEAP1 promoter. Downregulation of SELENBP1 is induced by miR-4786-3p binding to the 3' untranslated region (UTR) of SELENBP1. CONCLUSION SENENBP1 is identified as a novel protective biomarker for OSCC patients. Targeting at the miR-4786-3p-SELENBP1-KEAP1-NRF2 signaling axis may enhance the efficacy of chemotherapy for OSCC.
Collapse
Affiliation(s)
- Hui Zeng
- School of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Xubing Zhao
- Department of Emergency, Affiliated Stomatology Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Chengfang Tang
- School of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China.
| |
Collapse
|
13
|
Nayak V, Singh KRB, Singh AK, Singh RP. Potentialities of selenium nanoparticles in biomedical science. NEW J CHEM 2021. [DOI: 10.1039/d0nj05884j] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Selenium nanoparticles (SeNPs) have revolutionized biomedical domain and are still developing rapidly. Hence, this perspective elaborates SeNPs properties, synthesis, and biomedical applications, together with their potential for management of SARS-CoV-2.
Collapse
Affiliation(s)
- Vanya Nayak
- Department of Biotechnology
- Faculty of Science
- Indira Gandhi National Tribal University
- Amarkantak
- India
| | - Kshitij RB Singh
- Department of Chemistry
- Govt. V. Y. T. PG. Autonomous College
- Durg
- India
| | - Ajaya Kumar Singh
- Department of Chemistry
- Govt. V. Y. T. PG. Autonomous College
- Durg
- India
| | - Ravindra Pratap Singh
- Department of Biotechnology
- Faculty of Science
- Indira Gandhi National Tribal University
- Amarkantak
- India
| |
Collapse
|
14
|
Jia Y, Dai J, Zeng Z. Potential relationship between the selenoproteome and cancer. Mol Clin Oncol 2020; 13:83. [PMID: 33133596 PMCID: PMC7590431 DOI: 10.3892/mco.2020.2153] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
The role of the selenoproteome, which is the collection of all proteins containing selenium in an organism, in cancer development, growth and progression requires further investigation, due to the importance of selenium in both cancer and immune system function. Data about the selenoproteome, including its differential expression, single nucleotide variations, copy number variations, methylation, pathways and overall survival (OS) in five leading types of cancer are available from the GSCALite website. Subsequent to the analysis of these datasets, it was revealed that there was increased expression of GPX3 in stomach adenocarcinoma and lung squamous cell carcinoma, SELENOV in oesophageal carcinoma, GPX8 and GPX4 in colon adenocarcinoma, TXNRD1 and SEPHS1 in hepatocellular carcinoma and GPX8 in lung adenocarcinoma were associated with poor survival. Decreased gene expression of SELENOP was indicated in liver hepatocellular carcinoma and GPX3, and SELENOW, SELENOK, SELENBP1 and SECISBP2 in lung adenocarcinoma were associated with a poor prognosis. OS data suggested that hypermethylation of GPX4 in colon adenocarcinoma, GPX8 in lung squamous cell carcinoma, GPX1 in stomach adenocarcinoma and GPX3 in lung adenocarcinoma was associated with low survival, as is hypomethylation of GPX5 in lung adenocarcinoma. The selenoproteome is heterogeneous, especially in its effect on the OS of patients with cancer. The present study demonstrated that the roles of GPX4 in colon adenocarcinoma, SCLY and SELENOV in oesophageal carcinoma, SEPHS1 in liver hepatocellular carcinoma, SELENOK in lung cancer, as well as SELENOM and SELENOW in stomach adenocarcinoma requires further research. The present study may lead to the identification of novel biomarkers or potential therapeutic targets for use in the treatment of cancers, such as colon adenocarcinoma, oesophageal carcinoma, liver hepatocellular carcinoma, lung cancer and stomach adenocarcinoma.
Collapse
Affiliation(s)
- Yi Jia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Jie Dai
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zhu Zeng
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,School of Basic Medical Science, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
15
|
Lee YM, Kim S, Park RY, Kim YS. Hepatitis B Virus-X Downregulates Expression of Selenium Binding Protein 1. Viruses 2020; 12:v12050565. [PMID: 32443734 PMCID: PMC7291177 DOI: 10.3390/v12050565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Selenium binding protein 1 (SELENBP1) has been known to be reduced in various types cancer, and epigenetic change is shown to be likely to account for the reduction of SELNEBP1 expression. With cDNA microarray comparative analysis, we found that SELENBP1 is markedly decreased in hepatitis B virus-X (HBx)-expressing cells. To clarify the effect of HBx on SELENBP1 expression, we compared the expression levels of SELENBP1 mRNA and protein by semi-quantitative RT-PCR, Northern blot, and Western blot. As expected, SELENBP1 expression was shown to be reduced in cells expressing HBx, and reporter gene analysis showed that the SELENBP1 promoter is repressed by HBx. In addition, the stepwise deletion of 5′ flanking promoter sequences resulted in a gradual decrease in basal promoter activity and inhibition of SELENBP1 expression by HBx. Moreover, immunohistochemistry on tissue microarrays containing 60 pairs of human liver tissue showed decreased intensity of SELENBP1 in tumor tissues as compared with their matched non-tumor liver tissues. Taken together, our findings suggest that inhibition of SELENBP1 expression by HBx might act as one of the causes in the development of hepatocellular carcinoma caused by HBV infection.
Collapse
Affiliation(s)
- Young-Man Lee
- Dasan Undergraduate College, Ajou University, Suwon 16499, Korea;
| | - Soojin Kim
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34134, Korea;
| | - Ran-Young Park
- Department of Smart Food & Drugs, Inje University, Gimhae 50834, Korea;
| | - Yeon-Soo Kim
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34134, Korea;
- Correspondence: ; Tel.: +82-42-821-8631
| |
Collapse
|
16
|
Wang Y, Zhu W, Chen X, Wei G, Jiang G, Zhang G. Selenium-binding protein 1 transcriptionally activates p21 expression via p53-independent mechanism and its frequent reduction associates with poor prognosis in bladder cancer. J Transl Med 2020; 18:17. [PMID: 31918717 PMCID: PMC6953137 DOI: 10.1186/s12967-020-02211-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 01/03/2020] [Indexed: 01/31/2023] Open
Abstract
Background Recent studies have shown that selenium-binding protein 1 (SELENBP1) is significantly down-regulated in a variety of solid tumors. Nevertheless, the clinical relevance of SELENBP1 in human bladder cancer has not been described in any detail, and the molecular mechanism underlying its inhibitory role in cancer cell growth is largely unknown. Methods SELENBP1 expression levels in tumor tissues and adjacent normal tissues were evaluated using immunoblotting assay. The association of SELENBP1 expression, clinicopathological features, and clinical outcome was determined using publicly available dataset from The Cancer Genome Atlas bladder cancer (TCGA-BLCA) cohort. DNA methylation in SELENBP1 gene was assessed using online MEXPRESS tool. We generated stable SELENBP1-overexpression and their corresponding control cell lines to determine its potential effect on cell cycle and transcriptional activity of p21 by using flow cytometry and luciferase reporter assay, respectively. The dominant-negative mutant constructs, TAM67 and STAT1 Y701F, were employed to define the roles of c-Jun and STAT1 in the regulation of p21 protein. Results Here, we report that the reduction of SELENBP1 is a frequent event and significantly correlates with tumor progression as well as unfavorable prognosis in human bladder cancer. By utilizing TCGA-BLCA cohort, DNA hypermethylation, especially in gene body, is shown to be likely to account for the reduction of SELENBP1 expression. However, an apparent paradox is observed in its 3′-UTR region, in which DNA methylation is positively related to SELENBP1 expression. More importantly, we verify the growth inhibitory role for SELENBP1 in human bladder cancer, and further report a novel function for SELENBP1 in transcriptionally modulating p21 expression through a p53-independent mechanism. Instead, ectopic expression of SELENBP1 pronouncedly attenuates the phosphorylation of c-Jun and STAT1, both of which are indispensable for SELENBP1-mediated transcriptional induction of p21, thereby resulting in the G0/G1 phase cell cycle arrest in bladder cancer cell. Conclusions Taken together, our findings provide clinical and molecular insights into improved understanding of the tumor suppressive role for SELENBP1 in human bladder cancer, suggesting that SELENBP1 could potentially be utilized as a prognostic biomarker as well as a therapeutic target in future cancer therapy.
Collapse
Affiliation(s)
- Yulei Wang
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,School of Medicine, South China University of Technology, Guangzhou, 510641, China.
| | - Wenzhen Zhu
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xiaoqing Chen
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Guangnan Wei
- School of Medicine, South China University of Technology, Guangzhou, 510641, China
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guochun Zhang
- Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,School of Medicine, South China University of Technology, Guangzhou, 510641, China.
| |
Collapse
|
17
|
Nangraj AS, Selvaraj G, Kaliamurthi S, Kaushik AC, Cho WC, Wei DQ. Integrated PPI- and WGCNA-Retrieval of Hub Gene Signatures Shared Between Barrett's Esophagus and Esophageal Adenocarcinoma. Front Pharmacol 2020; 11:881. [PMID: 32903837 PMCID: PMC7438937 DOI: 10.3389/fphar.2020.00881] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/28/2020] [Indexed: 02/05/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a deadly cancer with high mortality rate, especially in economically advanced countries, while Barrett's esophagus (BE) is reported to be a precursor that strongly increases the risk of EAC. Due to the complexity of these diseases, their molecular mechanisms have not been revealed clearly. This study aims to explore the gene signatures shared between BE and EAC based on integrated network analysis. We obtained EAC- and BE-associated microarray datasets GSE26886, GSE1420, GSE37200, and GSE37203 from the Gene Expression Omnibus and ArrayExpress using systematic meta-analysis. These data were accompanied by clinical data and RNAseq data from The Cancer Genome Atlas (TCGA). Weighted gene co-expression network analysis (WGCNA) and differentially expressed gene (DEG) analysis were conducted to explore the relationship between gene sets and clinical traits as well as to discover the key relationships behind the co-expression modules. A differentially expressed gene-based protein-protein interaction (PPI) complex was used to extract hub genes through Cytoscape plugins. As a result, 403 DEGs were excavated, comprising 236 upregulated and 167 downregulated genes, which are involved in the cell cycle and replication pathways. Forty key genes were identified using modules of MCODE, CytoHubba, and CytoNCA with different algorithms. A dark-gray module with 207 genes was identified which having a high correlation with phenotype (gender) in the WGCNA. Furthermore, five shared hub gene signatures (SHGS), namely, pre-mRNA processing factor 4 (PRPF4), serine and arginine-rich splicing factor 1 (SRSF1), heterogeneous nuclear ribonucleoprotein M (HNRNPM), DExH-Box Helicase 9 (DHX9), and origin recognition complex subunit 2 (ORC2), were identified between BE and EAC. SHGS enrichment denotes that RNA metabolism and splicosomes play a key role in esophageal cancer development and progress. We conclude that the PPI complex and WGCNA co-expression network highlight the importance of phenotypic identifying hub gene signatures for BE and EAC.
Collapse
Affiliation(s)
- Asma Sindhoo Nangraj
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Gurudeeban Selvaraj
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
- *Correspondence: Gurudeeban Selvaraj, ; Dong Qing Wei,
| | - Satyavani Kaliamurthi
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
| | - Aman Chandra Kaushik
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Dong Qing Wei
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
- Peng Cheng Laboratory, Shenzhen, China
- *Correspondence: Gurudeeban Selvaraj, ; Dong Qing Wei,
| |
Collapse
|
18
|
DNA Methylation of miR-122 Aggravates Oxidative Stress in Colitis Targeting SELENBP1 Partially by p65NF- κB Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5294105. [PMID: 31019652 PMCID: PMC6451819 DOI: 10.1155/2019/5294105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/22/2018] [Accepted: 01/15/2019] [Indexed: 02/08/2023]
Abstract
Aberrant microRNA (miRNA) expressions contribute to the development and progression of various diseases, including Crohn's disease (CD). However, the accurate mechanisms of miRNAs in CD are definitely unclear. We employed colonic tissue samples from normal volunteers and CD patients, an acute mice colitis model induced by 2,4,6-trinitro-benzene-sulfonic acid (TNBS), and a cellular oxidative stress model induced by H2O2 in HT-29 cells to determine the effects of oxidative stress on expressions of miR-122, selenium-binding protein 1 (SELENBP1, SBP1), p65 nuclear factor κB (p65NF-κB) signaling, and DNA methylation. We found that SBP1 was mainly located on epithelial cells and was significantly increased in patients with active CD. SBP1 was the target gene of miR-122. miR-122 expression was downregulated while SBP1 expression was upregulated under TNBS-induced colitis or oxidative stress. Pre-miR-122 or siRNA SBP1 (si-SBP1) treatment ameliorated acute TNBS-induced colitis and H2O2-induced oxidative stress. Cotreatment of pre-miR-122 and si-SBP1 enhanced these effects. Besides, pre-miR-122 and si-SBP1 obviously activated the p65NF-κB signaling by phosphorylation of IκBα. Bisulfite sequencing of the CpG islands in the promoter region of miR-122 showed that CpG methylation was significantly increased under oxidative stress. Treating cells with 5′-AZA which was well known as a DNA-demethylating agent significantly increased miR-122 expression. Our results suggest that oxidative stress-induced DNA methylation of miR-122 aggravates colitis targeting SELENBP1 partially by p65NF-κB signaling and may promote the progression of CD.
Collapse
|
19
|
Wu J, Jin S, Gu W, Wan F, Zhang H, Shi G, Qu Y, Ye D. Construction and Validation of a 9-Gene Signature for Predicting Prognosis in Stage III Clear Cell Renal Cell Carcinoma. Front Oncol 2019; 9:152. [PMID: 30941304 PMCID: PMC6433707 DOI: 10.3389/fonc.2019.00152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose: Aim of this study was to develop a multi-gene signature to help better predict prognosis for stage III renal cell carcinoma (RCC) patients. Methods: Fourteen pairs of stage III tumor and normal tissues mRNA expression data from GSE53757 and 16 pairs mRNA expression data from TCGA clear cell RCC database were used to analyze differentially expressed genes between tumor and normal tissues. Common different expressed genes in both datasets were used for further modeling. Lasso Cox regression analysis was performed to select and build prognostic multi-gene signature in TCGA stage III kidney cancer patients (N = 122). Then, the multi-gene signature was validated in stage III renal cancer cases in Fudan University Shanghai Cancer Center (N = 77). C-index and time-dependent ROC were used to test the efficiency of this signature in predicting overall survival. Results: In total, 1,370 common different expressed genes were found between tumor and normal tissues in both datasets. After Lasso Cox modeling, nine mRNAs were finally identified to build a classifier. Using this classifier, we could classify stage III clear cell RCC patients into high-risk group and low-risk group. Prognosis was significantly different between these groups in discovery TCGA cohort, validation FUSCC cohort and entire set (All P < 0.001). Multivariate cox regression in entire set (N = 199) revealed that risk group classified by 9-gene signature, age of diagnosis, pN stage and ISUP grade were independent prognostic factor of overall survival in stage III kidney cancer patients. Conclusion: We developed a robust multi-gene classifier that can effectively classify stage III RCC patients into groups with low and high risk of poor prognosis. This signature may help select high-risk patients who require more aggressive adjuvant target therapy or immune therapy.
Collapse
Affiliation(s)
- Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weijie Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Tan HW, Mo HY, Lau ATY, Xu YM. Selenium Species: Current Status and Potentials in Cancer Prevention and Therapy. Int J Mol Sci 2018; 20:ijms20010075. [PMID: 30585189 PMCID: PMC6337524 DOI: 10.3390/ijms20010075] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/10/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Selenium (Se) acts as an essential trace element in the human body due to its unique biological functions, particularly in the oxidation-reduction system. Although several clinical trials indicated no significant benefit of Se in preventing cancer, researchers reported that some Se species exhibit superior anticancer properties. Therefore, a reassessment of the status of Se and Se compounds is necessary in order to provide clearer insights into the potentiality of Se in cancer prevention and therapy. In this review, we organize relevant forms of Se species based on the three main categories of Se-inorganic, organic, and Se-containing nanoparticles (SeNPs)-and overview their potential functions and applications in oncology. Here, we specifically focus on the SeNPs as they have tremendous potential in oncology and other fields. In general, to make better use of Se compounds in cancer prevention and therapy, extensive further study is still required to understand the underlying mechanisms of the Se compounds.
Collapse
Affiliation(s)
- Heng Wee Tan
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China.
| | - Hai-Ying Mo
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China.
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China.
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
21
|
Zhang XY, Gao PT, Yang X, Cai JB, Ding GY, Zhu XD, Ji Y, Shi GM, Shen YH, Zhou J, Fan J, Sun HC, Yang LX, Huang C. Reduced selenium-binding protein 1 correlates with a poor prognosis in intrahepatic cholangiocarcinoma and promotes the cell epithelial-mesenchymal transition. Am J Transl Res 2018; 10:3567-3578. [PMID: 30662608 PMCID: PMC6291736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/26/2018] [Indexed: 06/09/2023]
Abstract
Recent studies have found that selenium-binding protein 1 (SBP1) is downregulated in various malignant tumors. Nevertheless, the role of SBP1 in intrahepatic cholangiocarcinoma (ICC) is largely unknown. In the present study, we aimed to explore the clinical significance and biological function of SBP1 in ICC. Western blotting and immunohistochemistry were performed to evaluate SBP1 expression in ICC tissues, and correlations between SBP1 and clinicopathological parameters were further assessed. The prognostic significance of SBP1 in ICC patients was evaluated via Kaplan-Meier and Cox regression analyses. Moreover, we used RBE, a human ICC cell line, to study the effects of SBP1 knockdown on ICC cell proliferation, migration and invasion. Finally, the expression levels of epithelial-mesenchymal transition-related markers, including snail, vimentin, and E-cadherin, were investigated via Western blotting and immunohistochemistry. The results showed that SBP1 expression was significantly downregulated in ICC tumor tissues, especially in tumor tissues from ICC patients with recurrence or tumor vascular invasion, compared with that in peritumoral tissues (all P < 0.05). In addition, the reduction in SBP1 expression was related to microvascular invasion, lymphatic metastasis, and tumor-node-metastasis (TNM) stage (all P < 0.05). Furthermore, the SBP1 expression level was an independent prognostic factor in ICC (P < 0.05). Knockdown of SBP1 resulted in decreased in vitro proliferation, migration and invasion ability. Low SBP1 expression also resulted in the upregulation of mesenchymal markers such as vimentin and snail. In conclusion, SBP1 may be a prognostic indicator for patients with ICC as well as a potential target for ICC treatment.
Collapse
Affiliation(s)
- Xin-Yu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Ping-Ting Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Xuan Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jia-Bin Cai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Guang-Yu Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Guo-Ming Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Liu-Xiao Yang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| |
Collapse
|
22
|
Caswell DR, Chuang CH, Ma RK, Winters IP, Snyder EL, Winslow MM. Tumor Suppressor Activity of Selenbp1, a Direct Nkx2-1 Target, in Lung Adenocarcinoma. Mol Cancer Res 2018; 16:1737-1749. [PMID: 30002193 DOI: 10.1158/1541-7786.mcr-18-0392] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 12/18/2022]
Abstract
The Nkx2-1 transcription factor promotes differentiation of lung epithelial lineages and suppresses malignant progression of lung adenocarcinoma. However, targets of Nkx2-1 that limit tumor growth and progression remain incompletely understood. Here, direct Nkx2-1 targets are identified whose expression correlates with Nkx2-1 activity in human lung adenocarcinoma. Selenium-binding protein 1 (Selenbp1), an Nkx2-1 effector that limits phenotypes associated with lung cancer growth and metastasis, was investigated further. Loss- and gain-of-function approaches demonstrate that Nkx2-1 is required and sufficient for Selenbp1 expression in lung adenocarcinoma cells. Interestingly, Selenbp1 knockdown also reduced Nkx2-1 expression and Selenbp1 stabilized Nkx2-1 protein levels in a heterologous system, suggesting that these genes function in a positive feedback loop. Selenbp1 inhibits clonal growth and migration and suppresses growth of metastases in an in vivo transplant model. Genetic inactivation of Selenbp1, using CRISPR/Cas9, also enhanced primary tumor growth in autochthonous lung adenocarcinoma mouse models. Collectively, these data demonstrate that Selenbp1 is a direct target of Nkx2-1, which inhibits lung adenocarcinoma growth in vivo Implications: Selenbp1 is an important suppressor of lung tumor growth that functions in a positive feedback loop with Nkx2-1, and whose loss is associated with worse patient outcome. Mol Cancer Res; 16(11); 1737-49. ©2018 AACR.
Collapse
Affiliation(s)
- Deborah R Caswell
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California
| | - Chen-Hua Chuang
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Rosanna K Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ian P Winters
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Eric L Snyder
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Monte M Winslow
- Cancer Biology Program, Stanford University School of Medicine, Stanford, California. .,Department of Genetics, Stanford University School of Medicine, Stanford, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
23
|
Gao PT, Ding GY, Yang X, Dong RZ, Hu B, Zhu XD, Cai JB, Ji Y, Shi GM, Shen YH, Zhou J, Fan J, Sun HC, Huang C. Invasive potential of hepatocellular carcinoma is enhanced by loss of selenium-binding protein 1 and subsequent upregulation of CXCR4. Am J Cancer Res 2018; 8:1040-1049. [PMID: 30034941 PMCID: PMC6048402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/08/2018] [Indexed: 06/08/2023] Open
Abstract
Decreased selenium-binding protein 1 (SBP1) is associated with increased invasion and poor prognosis of hepatocellular carcinoma (HCC). However, the underlying mechanism remains unknown. To unravel this mechanism, HCC cells expressing SBP1 were constructed and the impact on migration, invasion, and epithelial-mesenchymal transition (EMT) was evaluated. SBP1 expression reduced HCC cell migration and invasion by inhibiting EMT. Gene expression profiles of control and SBP1 expressing HCC cells revealed 186 differentially expressed genes, of which fibroblast growth factor 5, vascular endothelial growth factor receptor 1, and C-X-C motif chemokine receptor 4 (CXCR4) showed the greatest differences. CXCR4 expression was inhibited by SBP1 and restored the migration and invasion ability of HCC cells through activation of AKT signaling. Tumor samples from 200 HCC patients supported our in vitro findings and revealed an inverse correlation between SBP1 and CXCR4 expression. Patients with low SBP1 and high CXCR4 expression had the poorest prognosis and survival rate. Our results suggest that downregulation of SBP1 induces increased CXCR4 expression and results in EMT of HCC cells. Together, SBP1 and CXCR4 are promising potential biomarkers and therapeutic targets for HCC patients.
Collapse
Affiliation(s)
- Ping-Ting Gao
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Guang-Yu Ding
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Xuan Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Rui-Zhao Dong
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Bo Hu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jia-Bin Cai
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Guo-Ming Shi
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| | - Cheng Huang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan UniversityShanghai, China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of EducationShanghai, China
| |
Collapse
|
24
|
Lee KTW, Smith RA, Gopalan V, Lam AK. Targeted Single Gene Mutation in Esophageal Adenocarcinoma. Methods Mol Biol 2018; 1756:213-229. [PMID: 29600373 DOI: 10.1007/978-1-4939-7734-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Esophageal adenocarcinoma is heterogeneous and studies have reviewed many important mutations that contribute to the pathogenesis of the cancer. These discoveries have helped paved the way into identifying new gene markers or gene targets to develop novel molecular directed therapy for better patient outcomes in esophageal adenocarcinoma. Despite the recent bloom in next-generation sequencing, Sanger sequencing still represents the gold standard method for the study of the driver genes in esophageal adenocarcinoma. This chapter focuses on the sequencing techniques in identification of single gene mutations.
Collapse
Affiliation(s)
- Katherine T W Lee
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| | - Robert A Smith
- Genomics Research Centre, School of Biomedical Science, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia.
| |
Collapse
|
25
|
Somatic DNA Copy-Number Alterations Detection for Esophageal Adenocarcinoma Using Digital Polymerase Chain Reaction. Methods Mol Biol 2018. [PMID: 29600372 DOI: 10.1007/978-1-4939-7734-5_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Somatic copy-number alterations are commonly found in cancer and play key roles in activating oncogenes and deactivating tumor suppressor genes. Digital polymerase chain reaction is an effective way to detect the changes in copy number. In esophageal adenocarcinoma, detection of somatic copy-number alterations could predict the prognosis of patients as well as the response to therapy. This chapter will review the methods involved in digital polymerase chain reaction for the research or potential clinical applications in esophageal adenocarcinoma.
Collapse
|
26
|
Ferrer-Torres D, Nancarrow DJ, Kuick R, Thomas DG, Nadal E, Lin J, Chang AC, Reddy RM, Orringer MB, Taylor JMG, Wang TD, Beer DG. Genomic similarity between gastroesophageal junction and esophageal Barrett's adenocarcinomas. Oncotarget 2018; 7:54867-54882. [PMID: 27363029 PMCID: PMC5342387 DOI: 10.18632/oncotarget.10253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
The current high mortality rate of esophageal adenocarcinoma (EAC) reflects frequent presentation at an advanced stage. Recent efforts utilizing fluorescent peptides have identified overexpressed cell surface targets for endoscopic detection of early stage Barrett's-derived EAC. Unfortunately, 30% of EAC patients present with gastroesophageal junction adenocarcinomas (GEJAC) and lack premalignant Barrett's metaplasia, limiting this early detection strategy. We compared mRNA profiles from 52 EACs (tubular EAC; tEAC) collected above the gastroesophageal junction with 70 GEJACs, 8 normal esophageal and 5 normal gastric mucosa samples. We also analyzed our previously published whole-exome sequencing data in a large cohort of these tumors. Principal component analysis, hierarchical clustering and survival-based analyses demonstrated that GEJAC and tEAC were highly similar, with only modest differences in expression and mutation profiles. The combined expression cohort allowed identification of 49 genes coding cell surface targets overexpressed in both GEJAC and tEAC. We confirmed that three of these candidates (CDH11, ICAM1 and CLDN3) were overexpressed in tumors when compared to normal esophagus, normal gastric and non-dysplastic Barrett's, and localized to the surface of tumor cells. Molecular profiling of tEAC and GEJAC tumors indicated extensive similarity and related molecular processes. Identified genes that encode cell surface proteins overexpressed in both Barrett's-derived EAC and those that arise without Barrett's metaplasia will allow simultaneous detection strategies.
Collapse
Affiliation(s)
- Daysha Ferrer-Torres
- Cancer Biology, Program in Biomedical Science, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Derek J Nancarrow
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rork Kuick
- Center for Cancer Biostatistics, Department of Biostatistics, School of Public Health, Ann Arbor, Michigan, USA
| | - Dafydd G Thomas
- Department of Pathology and Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ernest Nadal
- Medical Oncology Department, Catalan Institute of Oncology, Barcelona, Spain
| | - Jules Lin
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Andrew C Chang
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rishindra M Reddy
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mark B Orringer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jeremy M G Taylor
- Center for Cancer Biostatistics, Department of Biostatistics, School of Public Health, Ann Arbor, Michigan, USA
| | - Thomas D Wang
- Department of Medicine and Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David G Beer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Schott M, de Jel MM, Engelmann JC, Renner P, Geissler EK, Bosserhoff AK, Kuphal S. Selenium-binding protein 1 is down-regulated in malignant melanoma. Oncotarget 2018. [PMID: 29535818 PMCID: PMC5828193 DOI: 10.18632/oncotarget.23853] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Selenium-binding protein 1 (SELENBP1) expression is reduced in various epithelial cancer entities compared to corresponding normal tissue and has already been described as a tumor suppressor involved in the regulation of cell proliferation, senescence, migration and apoptosis. We identified SELENBP1 to be down-regulated in cutaneous melanoma, a malignant cancer of pigment-producing melanocytes in the skin, which leads to the assumption that SELENBP1 also functions as tumor suppressor in the skin, as shown by others e.g. for prostate or lung carcinoma. However, in vitro analyses indicate that SELENBP1 re-expression in human melanoma cell lines has no impact on cell proliferation, migration or tube formation of the tumor cells themselves when compared to control-transfected cells. Interestingly, supernatant taken from melanoma cell lines transfected with a SELENBP1 re-expression plasmid led to suppression of vessel formation of HMEC cells. Furthermore, SELENBP1 re-expression alters the sensitivity of melanoma cells for Vemurafenib treatment. The data also hint to a functional interaction of SELENBP1 with GPX1 (Glutathione peroxidase 1). Low SELENBP1 mRNA levels correlate inversely with GPX1 expression in melanoma. The re-expression of SELENBP1 combined with down-regulation of GPX1 expression led to reduction of the proliferation of melanoma cells. In summary, SELENBP1 influences the tumor microenvironment and SELENBP1 action is functionally influenced by GPX1.
Collapse
Affiliation(s)
- Mandy Schott
- University of Erlangen, Institute of Biochemistry, Biochemistry and Molecular Medicine, Erlangen, Germany
| | - Miriam M de Jel
- University of Erlangen, Institute of Biochemistry, Biochemistry and Molecular Medicine, Erlangen, Germany
| | - Julia C Engelmann
- University of Regensburg, Institute of Functional Genomics, Statistical Bioinformatics, Regensburg, Germany
| | - Philipp Renner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Anja K Bosserhoff
- University of Erlangen, Institute of Biochemistry, Biochemistry and Molecular Medicine, Erlangen, Germany
| | - Silke Kuphal
- University of Erlangen, Institute of Biochemistry, Biochemistry and Molecular Medicine, Erlangen, Germany
| |
Collapse
|
28
|
Bao Y, Wang Q, Guo Y, Chen Z, Li K, Yang Y, Zhang H, Dong H, Shen K, Yang W. PRSS8 methylation and its significance in esophageal squamous cell carcinoma. Oncotarget 2017; 7:28540-55. [PMID: 27081034 PMCID: PMC5053744 DOI: 10.18632/oncotarget.8677] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022] Open
Abstract
Esophageal cancer is one of the most common cancers worldwide, and the incidence and mortality is increasing rapidly in recent years in China, but the underlying mechanisms are largely unclear. Herein we found that the expression of PRSS8, a serine protease prostasin, is significantly decreased in esophageal squamous cell carcinomas (ESCC) at mRNA and protein levels. The reduction of PRSS8 was well correlated with poor differentiation and shorter survival time. Interestingly, ESCC stromal expression of PRSS8 was significantly correlated with stromal lymphocyte infiltration and cancer progression. Methylation specific PCR showed that PRSS8 was hypermethylated in ESCC tissues and ESCC cell lines, which was linked to the downregulation of PRSS8 expression and decreased activities of PRSS8 promoter. De-methylation agent decitabine was able to restore PRSS8 expression, leading to the inhibition of cancer cell proliferation, motility, migration and cell cycle arrest. However, the restored PRSS8 and its tumor inhibition could be reversed by small interfering RNA targeting PRSS8. Mechanistic study showed that tumor inhibition of PRSS8 may be associated with proliferation- and epithelial mesenchymal transition - related proteins in ESCC cells. In conclusion, our finding showed that PRSS8 methylation and its stromal expression had important clinical significance in ESCC.
Collapse
Affiliation(s)
- Yonghua Bao
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Qian Wang
- Department of Immunology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yongchen Guo
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Zhiguo Chen
- Department of Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Kai Li
- Department of Pathology, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang 453003, China
| | - Yiqiong Yang
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China
| | - Huijuan Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang 453003, China
| | - Huali Dong
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Kui Shen
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wancai Yang
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining 272067, China.,Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
29
|
Maag JLV, Fisher OM, Levert-Mignon A, Kaczorowski DC, Thomas ML, Hussey DJ, Watson DI, Wettstein A, Bobryshev YV, Edwards M, Dinger ME, Lord RV. Novel Aberrations Uncovered in Barrett's Esophagus and Esophageal Adenocarcinoma Using Whole Transcriptome Sequencing. Mol Cancer Res 2017; 15:1558-1569. [PMID: 28751461 DOI: 10.1158/1541-7786.mcr-17-0332] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 06/30/2017] [Accepted: 07/21/2017] [Indexed: 11/16/2022]
Abstract
Esophageal adenocarcinoma (EAC) has one of the fastest increases in incidence of any cancer, along with poor five-year survival rates. Barrett's esophagus (BE) is the main risk factor for EAC; however, the mechanisms driving EAC development remain poorly understood. Here, transcriptomic profiling was performed using RNA-sequencing (RNA-seq) on premalignant and malignant Barrett's tissues to better understand this disease. Machine-learning and network analysis methods were applied to discover novel driver genes for EAC development. Identified gene expression signatures for the distinction of EAC from BE were validated in separate datasets. An extensive analysis of the noncoding RNA (ncRNA) landscape was performed to determine the involvement of novel transcriptomic elements in Barrett's disease and EAC. Finally, transcriptomic mutational investigation of genes that are recurrently mutated in EAC was performed. Through these approaches, novel driver genes were discovered for EAC, which involved key cell cycle and DNA repair genes, such as BRCA1 and PRKDC. A novel 4-gene signature (CTSL, COL17A1, KLF4, and E2F3) was identified, externally validated, and shown to provide excellent distinction of EAC from BE. Furthermore, expression changes were observed in 685 long noncoding RNAs (lncRNA) and a systematic dysregulation of repeat elements across different stages of Barrett's disease, with wide-ranging downregulation of Alu elements in EAC. Mutational investigation revealed distinct pathways activated between EAC tissues with or without TP53 mutations compared with Barrett's disease. In summary, transcriptome sequencing revealed altered expression of numerous novel elements, processes, and networks in EAC and premalignant BE.Implications: This study identified opportunities to improve early detection and treatment of patients with BE and esophageal adenocarcinoma. Mol Cancer Res; 15(11); 1558-69. ©2017 AACR.
Collapse
Affiliation(s)
- Jesper L V Maag
- Genome Informatics, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Oliver M Fisher
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - Angelique Levert-Mignon
- Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - Dominik C Kaczorowski
- Genome Informatics, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Melissa L Thomas
- Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia.,University of Notre Dame School of Medicine, Sydney, Australia
| | - Damian J Hussey
- Department of Surgery, Flinders University, Adelaide, Australia
| | - David I Watson
- Department of Surgery, Flinders University, Adelaide, Australia
| | - Antony Wettstein
- Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - Yuri V Bobryshev
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - Melanie Edwards
- Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia.,University of Notre Dame School of Medicine, Sydney, Australia
| | - Marcel E Dinger
- Genome Informatics, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, Australia. .,Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Reginald V Lord
- Gastroesophageal Cancer Program, St. Vincent's Centre for Applied Medical Research, Sydney, Australia. .,University of Notre Dame School of Medicine, Sydney, Australia
| |
Collapse
|
30
|
Yu-Rice Y, Edassery SL, Urban N, Hellstrom I, Hellstrom KE, Deng Y, Li Y, Luborsky JL. Selenium-Binding Protein 1 (SBP1) autoantibodies in ovarian disorders and ovarian cancer. Reproduction 2016; 153:277-284. [PMID: 27965399 DOI: 10.1530/rep-16-0265] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 12/30/2022]
Abstract
Infertility is a risk factor for ovarian cancer (OvCa). The goal was to determine if antibodies to selenium-binding protein 1 (SBP1), an autoantibody we identified in patients with premature ovarian failure (POF), occurs in both infertility and OvCa patients, and thus could be associated with preneoplasia. Anti-SBP1 was measured by immunoassay against recombinant SBP1, in sera from OvCa (n = 41), infertility (n = 92) and control (n = 87) patients. Infertility causes were POF, unexplained, irregular ovulation or endometriosis. The percent of anti-SBP1-positive sera was higher in POF (P = 0.02), irregular ovulation (P = 0.001), unexplained causes (P = 0.02), late (III-IV)-stage OvCa (P = 0.02) but was not significant in endometriosis, benign ovarian tumors/cysts, early stage (I-II) OvCa or uterine cancer compared to healthy controls. Anti-SBP1 was significantly higher in women with serous (P = 0.04) but not non-serous (P = 0.33) OvCa compared to controls. Also, we determined if anti-SBP1 was associated with CA125 or anti-TP53, markers often studied in OvCa. Anti-TP53 and CA125 were measured by established immunoassays. The ability of anti-SBP1 alone to discriminate infertility or OvCa from controls or when combined with anti-TP53 and CA125, to identify OvCa was evaluated by comparing the area under the curve (AUC) in ROC analysis. Anti-SBP1 alone discriminated infertility (AUC = 0.7; P = 0.001) or OvCa (AUC = 0.67; P = 0.03) from controls. The sensitivity and specificity of OvCa identification was increased by combining CA125, anti-TP53 and anti-SBP1 (AUC = 0.96). Therefore, anti-SBP1 occurs in infertile women with POF, ovulatory disturbances or unexplained infertility and in serous OvCa. This suggests an autoimmune process is associated with the development of serous OvCa.
Collapse
Affiliation(s)
- Yi Yu-Rice
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| | - Seby L Edassery
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| | - Nicole Urban
- Fred Hutchinson Cancer Research CenterSeattle, Washington, USA
| | - Ingegerd Hellstrom
- Department of PathologyHarborview Medical Center, University of Washington, Seattle, Washington, USA
| | - Karl Erik Hellstrom
- Department of PathologyHarborview Medical Center, University of Washington, Seattle, Washington, USA
| | - Youping Deng
- Department of Bioinformatics and BiostatisticsRush University Medical Center, Chicago, Illinois, USA
| | - Yan Li
- Department of Bioinformatics and BiostatisticsRush University Medical Center, Chicago, Illinois, USA
| | - Judith L Luborsky
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
31
|
Duggan SP, Behan FM, Kirca M, Zaheer A, McGarrigle SA, Reynolds JV, Vaz GMF, Senge MO, Kelleher D. The characterization of an intestine-like genomic signature maintained during Barrett's-associated adenocarcinogenesis reveals an NR5A2-mediated promotion of cancer cell survival. Sci Rep 2016; 6:32638. [PMID: 27586588 PMCID: PMC5009315 DOI: 10.1038/srep32638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
Barrett’s oesophagus (BO), an intestinal-type metaplasia (IM), typically arising in conjunction with gastro-oesophageal reflux disease, is a prominent risk factor for the development of oesophageal adenocarcinoma (OAC). The molecular similarities between IM and normal intestinal tissues are ill-defined. Consequently, the contribution of intestine-enriched factors expressed within BO to oncogenesis is unclear. Herein, using transcriptomics we define the intestine-enriched genes expressed in meta-profiles of BO and OAC. Interestingly, 77% of the genes differentially expressed in a meta-profile of BO were similarly expressed in intestinal tissues. Furthermore, 85% of this intestine-like signature was maintained upon transition to OAC. Gene networking analysis of transcription factors within this signature revealed a network centred upon NR5A2, GATA6 and FOXA2, whose over-expression was determined in a cohort of BO and OAC patients. Simulated acid reflux was observed to induce the expression of both NR5A2 and GATA6. Using siRNA-mediated silencing and an NR5A2 antagonist we demonstrate that NR5A2-mediated cancer cell survival is facilitated through augmentation of GATA6 and anti-apoptotic factor BCL-XL levels. Abrogation of NR5A2-GATA6 expression in conjunction with BCL-XL co-silencing resulted in synergistically increased sensitivity to chemotherapeutics and photo-dynamic therapeutics. These findings characterize the intestine-like signature associated with IM which may have important consequences to adenocarcinogenesis.
Collapse
Affiliation(s)
- Shane P Duggan
- Department of Medicine, Division of Gastroenterology, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia, Canada.,Life Science Institute, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada
| | - Fiona M Behan
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, St James' Hospital, Dublin, Ireland
| | - Murat Kirca
- Department of Gastroenterology, St James' Hospital, Dublin, Ireland
| | - Abdul Zaheer
- Department of Gastroenterology, St James' Hospital, Dublin, Ireland
| | - Sarah A McGarrigle
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St James' Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St James' Hospital, Dublin 8, Ireland
| | - Gisela M F Vaz
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity College Dublin, the University of Dublin, St James' Hospital, Dublin 8, Ireland
| | - Mathias O Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity College Dublin, the University of Dublin, St James' Hospital, Dublin 8, Ireland
| | - Dermot Kelleher
- Department of Medicine, Division of Gastroenterology, University of British Columbia, 2775 Laurel Street, Vancouver, British Columbia, Canada.,Life Science Institute, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Chen F, Chen C, Qu Y, Xiang H, Ai Q, Yang F, Tan X, Zhou Y, Jiang G, Zhang Z. Selenium-binding protein 1 in head and neck cancer is low-expression and associates with the prognosis of nasopharyngeal carcinoma. Medicine (Baltimore) 2016; 95:e4592. [PMID: 27583873 PMCID: PMC5008557 DOI: 10.1097/md.0000000000004592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Selenium-binding protein 1 (SELENBP1) expression is reduced markedly in many types of cancers and low SELENBP1 expression levels are associated with poor patient prognosis. METHODS SELENBP1 gene expression in head and neck squamous cell carcinoma (HNSCC) was analyzed with GEO dataset and characteristics of SELENBP1 expression in paraffin embedded tissue were summarized. Expression of SELENBP1 in nasopharyngeal carcinoma (NPC), laryngeal cancer, oral cancer, tonsil cancer, hypopharyngeal cancer and normal tissues were detected using immunohistochemistry, at last, 99 NPC patients were followed up more than 5 years and were analyzed the prognostic significance of SELENBP1. RESULTS Analysis of GEO dataset concluded that SELENBP1 gene expression in HNSCC was lower than that in normal tissue (P < 0.01), but there was no significant difference of SELENBP1 gene expression in different T-stage and N-stage (P > 0.05). Analysis of pathological section concluded that SELENBP1 in the majority of HNSCC is low expression and in cancer nests is lower expression than surrounding normal tissue, even associated with the malignant degree of tumor. Further study indicated the low SELENBP1 expression group of patients with NPC accompanied by poor overall survival and has significantly different comparing with the high expression group. CONCLUSION SELENBP1 expression was down-regulated in HNSCC, but has no associated with T-stage and N-stage of tumor. Low expression of SELENBP1 in patients with NPC has poor over survival, so SELENBP1 could be a novel biomarker for predicting prognosis.
Collapse
Affiliation(s)
- Fasheng Chen
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Chen Chen
- Research institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Yangang Qu
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Hua Xiang
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Qingxiu Ai
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Fei Yang
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Xueping Tan
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Yi Zhou
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
| | - Guang Jiang
- Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu Province, China
| | - Zixiong Zhang
- Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province
- Correspondence: Zixiong Zhang, Department of Otolaryngology Head and Neck Surgery, Central Hospital of Enshi Autonomous Prefecture, Enshi Autonomous Prefecture, Hubei Province 445000, PR China (e-mail: )
| |
Collapse
|
33
|
Myers AL, Lin L, Nancarrow DJ, Wang Z, Ferrer-Torres D, Thomas DG, Orringer MB, Lin J, Reddy RM, Beer DG, Chang AC. IGFBP2 modulates the chemoresistant phenotype in esophageal adenocarcinoma. Oncotarget 2016; 6:25897-916. [PMID: 26317790 PMCID: PMC4694874 DOI: 10.18632/oncotarget.4532] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 07/06/2015] [Indexed: 12/21/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) patients commonly present with advanced stage disease and demonstrate resistance to therapy, with response rates below 40%. Understanding the molecular mechanisms of resistance is crucial for improvement of clinical outcomes. IGFBP2 is a member of the IGFBP family of proteins that has been reported to modulate both IGF and integrin signaling and is a mediator of cell growth, invasion and resistance in other tumor types. In this study, high IGFBP2 expression was observed in a subset of primary EACs and was found to be significantly higher in patients with shorter disease-free intervals as well as in treatment-resistant EACs as compared to chemonaive EACs. Modulation of IGFBP2 expression in EAC cell lines promoted cell proliferation, migration and invasion, implicating a role in the metastatic potential of these cells. Additionally, knockdown of IGFBP2 sensitized EAC cells to cisplatin in a serum-dependent manner. Further in vitro exploration into this chemosensitization implicated both the AKT and ERK pathways. Silencing of IGFBP2 enhanced IGF1-induced immediate activation of AKT and reduced cisplatin-induced ERK activation. Addition of MEK1/2 (selumetinib or trametinib) or AKT (AKT Inhibitor VIII) inhibitors enhanced siIGFBP2-induced sensitization of EAC cells to cisplatin. These results suggest that targeted inhibition of IGFBP2 alone or together with either the MAPK or PI3K/AKT signaling pathway in IGFBP2-overexpressing EAC tumors may be an effective approach for sensitizing resistant EACs to standard neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Amy L Myers
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lin Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Zhuwen Wang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Dafydd G Thomas
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mark B Orringer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jules Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - David G Beer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Andrew C Chang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Zhao C, Zeng H, Wu RTY, Cheng WH. Loss of Selenium-Binding Protein 1 Decreases Sensitivity to Clastogens and Intracellular Selenium Content in HeLa Cells. PLoS One 2016; 11:e0158650. [PMID: 27404728 PMCID: PMC4942091 DOI: 10.1371/journal.pone.0158650] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/20/2016] [Indexed: 12/27/2022] Open
Abstract
Selenium-binding protein 1 (SBP1) is not a selenoprotein but structurally binds selenium. Loss of SBP1 during carcinogenesis usually predicts poor prognosis. Because genome instability is a hallmark of cancer, we hypothesize that SBP1 sequesters cellular selenium and sensitizes cancer cells to DNA-damaging agents. To test this hypothesis, we knocked down SBP1 expression in HeLa cervical cancer cells by employing a short hairpin RNA (shRNA) approach. Reduced sensitivity to hydrogen peroxide, paraquat and camptothecin, reactive oxygen species content, and intracellular retention of selenium after selenomethionine treatment were observed in SBP1 shRNA HeLa cells. Results from Western analyses showed that treatment of HeLa cells with selenomethionine resulted in increased SBP1 protein expression in a dose-dependent manner. Knockdown of SBP1 rendered HeLa cells increased expression of glutathione peroxidase-1 but not glutathione peroxidase-4 protein levels and accelerated migration from a wound. Altogether, SBP1 retains supplemental selenium and sensitizes HeLa cancer cells to clastogens, suggesting a new cancer treatment strategy by sequestering selenium through SBP1.
Collapse
Affiliation(s)
- Changhui Zhao
- Department of Food Quality and Safety, College of Food Science and Engineering, Jilin University, Changchun, Jilin Province, 130062, China
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland 20742, United States of America
| | - Huawei Zeng
- United States Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota 58202, United States of America
| | - Ryan T. Y. Wu
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland 20742, United States of America
| | - Wen-Hsing Cheng
- Department of Food Science, Nutrition & Health Promotion, Mississippi State University, Mississippi State, Mississippi 39762, United States of America
| |
Collapse
|
35
|
Jeyapalan JN, Doctor GT, Jones TA, Alberman SN, Tep A, Haria CM, Schwalbe EC, Morley ICF, Hill AA, LeCain M, Ottaviani D, Clifford SC, Qaddoumi I, Tatevossian RG, Ellison DW, Sheer D. DNA methylation analysis of paediatric low-grade astrocytomas identifies a tumour-specific hypomethylation signature in pilocytic astrocytomas. Acta Neuropathol Commun 2016; 4:54. [PMID: 27229157 PMCID: PMC4882864 DOI: 10.1186/s40478-016-0323-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/04/2016] [Indexed: 12/30/2022] Open
Abstract
Low-grade gliomas (LGGs) account for about a third of all brain tumours in children. We conducted a detailed study of DNA methylation and gene expression to improve our understanding of the biology of pilocytic and diffuse astrocytomas. Pilocytic astrocytomas were found to have a distinctive signature at 315 CpG sites, of which 312 were hypomethylated and 3 were hypermethylated. Genomic analysis revealed that 182 of these sites are within annotated enhancers. The signature was not present in diffuse astrocytomas, or in published profiles of other brain tumours and normal brain tissue. The AP-1 transcription factor was predicted to bind within 200 bp of a subset of the 315 differentially methylated CpG sites; the AP-1 factors, FOS and FOSL1 were found to be up-regulated in pilocytic astrocytomas. We also analysed splice variants of the AP-1 target gene, CCND1, which encodes cell cycle regulator cyclin D1. CCND1a was found to be highly expressed in both pilocytic and diffuse astrocytomas, but diffuse astrocytomas have far higher expression of the oncogenic variant, CCND1b. These findings highlight novel genetic and epigenetic differences between pilocytic and diffuse astrocytoma, in addition to well-described alterations involving BRAF, MYB and FGFR1.
Collapse
Affiliation(s)
- Jennie N Jeyapalan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Gabriel T Doctor
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Tania A Jones
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Samuel N Alberman
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Alexander Tep
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Chirag M Haria
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Edward C Schwalbe
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Isabel C F Morley
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Alfred A Hill
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Magdalena LeCain
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Diego Ottaviani
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Steven C Clifford
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Ibrahim Qaddoumi
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ruth G Tatevossian
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, 38105-3678, USA
| | - David W Ellison
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, 38105-3678, USA.
| | - Denise Sheer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK.
| |
Collapse
|
36
|
Xie W, Yang M, Chan J, Sun T, Mucci LA, Penney KL, Lee GSM, Kantoff PW. Association of genetic variations of selenoprotein genes, plasma selenium levels, and prostate cancer aggressiveness at diagnosis. Prostate 2016; 76:691-9. [PMID: 26847995 PMCID: PMC5510241 DOI: 10.1002/pros.23160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/14/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Genetic variations in some of the selenoprotein genes, alone or together with an individual's selenium status, may influence risk or progression of prostate cancer. We investigated the impact of genetic variants of selenoproteins on plasma selenium levels and cancer aggressiveness at diagnosis in men with localized prostate cancer (PCa). METHODS The study cohort comprised 722 patients seen at Dana-Farber Cancer Institute who had localized/locally advanced PCa (i.e., stage T3 or less, N0, and M0) from 1994 to 2001. Fifty-five tagging single nucleotide polymorphisms (SNPs) from six selenoprotein genes (TXNRD1, TXNRD2, SEP15, GPX3, SELENBP1, and SEPP1) were analyzed. Logistic regression is used to examine associations of genotypes and plasma selenium levels with risk of aggressive disease, defined as D'Amico intermediate/high risk categories. Step down permutation was applied to adjust for multiple comparisons. RESULTS Three hundred and forty-eight patients (48%) had aggressive disease at diagnosis. Two SNPs were associated with cancer aggressiveness at diagnosis (unadjusted P = 0.017 and 0.018, respectively). The odds ratio for aggressive disease in patients carrying TXNRD2 rs1005873-AG/GG genotypes or SELENBP1 rs10788804-AG/AA genotypes was 1.54 (95% CI = 1.08, 2.20) and 1.45 (95% CI = 1.07, 1.98), respectively, compared to TXNRD2 rs1005873-AA or SELENBP1 rs10788804-GG carriers. Four SNPs in TXNRD2 (rs1005873, rs13054371, rs3788310, and rs9606174) and the rs230820 in SEPP1 were associated with plasma selenium levels (unadjusted P < 0.05). Permutation adjusted P-values were not statistically significant for all these comparisons at the cut-off point of 0.05. CONCLUSION We identified polymorphisms in selenoproteins that may influence the plasma selenium levels and may be associated with the risk of presenting with aggressive PCa in men with localized or locally advanced PCa. These results should be validated in other independent datasets.
Collapse
Affiliation(s)
- Wanling Xie
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Ming Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - June Chan
- Department of Epidemiology and Biostatistics and Urology, University of California, San Francisco, CA
| | - Tong Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Philip W. Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| |
Collapse
|
37
|
Tan X, Liao L, Wan YP, Li MX, Chen SH, Mo WJ, Zhao QL, Huang LF, Zeng GQ. Downregulation of selenium-binding protein 1 is associated with poor prognosis in lung squamous cell carcinoma. World J Surg Oncol 2016; 14:70. [PMID: 26956891 PMCID: PMC4782367 DOI: 10.1186/s12957-016-0832-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 03/01/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND We found that selenium-binding protein 1 (SBP1) was progressively decreased in the human bronchial epithelial carcinogenic processes. Knockdown of SBP1 in immortalized human bronchial epithelial cell line 16HBE cells significantly increased the efficiency of B[a]P-induced cell transformation. However, the relationship between SBP1 expression and clinicopathological factors of patients has not been defined completely. The specific role of SBP1 in prognosis of lung squamous cell carcinoma (LSCC) is still unknown. METHODS Tissue samples from 82 patients treated by pulmonary lobectomy for LSCC were used. Immunohistochemistry and western blotting were used to detect the expressions of SBP1 protein. The relationships between the expression level of SBP1 and the clinicopathological features of patients were analyzed. Cox proportional hazard regression analysis and Kaplan-Meier method were used to perform survival analysis. RESULTS Expressions of SBP1 proteins were significantly lower in LSCC tissues than that in the corresponding normal bronchial epithelium (NBE) tissues (P = 0.000). In LSCC, The expression levels of SBP1 had not correlated with patients' age, gender, smoking state, primary tumor stages (T), TNM clinical stages, and distant metastasis (M) (P > 0.05). However, downregulation of SBP1 was significantly associated with higher lymph node metastasis and lower overall survival rate (P < 0.05). Cox regression analysis indicated low expressions of SBP1 can be an independent prognostic factor for poor overall survival in LSCC patients (P = 0.002). CONCLUSIONS Downregulation of SBP1 may play a key role in the tumorigenic process of LSCC. SBP1 may be a novel potential prognostic factor of LSCC.
Collapse
Affiliation(s)
- Xing Tan
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Li Liao
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Yan-Ping Wan
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Mei-Xiang Li
- School of Medicine, University of South China, Hengyang, 421001, China.
| | - Si-Han Chen
- School of Medicine, University of South China, Hengyang, 421001, China.
| | - Wen-Juan Mo
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Qiong-Lan Zhao
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Li-Fang Huang
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| | - Gu-Qing Zeng
- School of Nursing, University of South China, 28# Changsheng Road West, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Gong J, Li L. Sodium Selenite Inhibits Proliferation of Gastric Cancer Cells by Inducing SBP1 Expression. TOHOKU J EXP MED 2016; 239:279-85. [DOI: 10.1620/tjem.239.279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jianzhuang Gong
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University
| | - Li Li
- Department of Blood Transfusion, Zhengzhou Central Hospital Affiliated to Zhengzhou University
| |
Collapse
|
39
|
Li B, Xu WW, Guan XY, Qin YR, Law S, Lee NPY, Chan KT, Tam PY, Li YY, Chan KW, Yuen HF, Tsao SW, He QY, Cheung ALM. Competitive Binding Between Id1 and E2F1 to Cdc20 Regulates E2F1 Degradation and Thymidylate Synthase Expression to Promote Esophageal Cancer Chemoresistance. Clin Cancer Res 2015; 22:1243-55. [PMID: 26475334 DOI: 10.1158/1078-0432.ccr-15-1196] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/15/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Chemoresistance is a major obstacle in cancer therapy. We found that fluorouracil (5-FU)-resistant esophageal squamous cell carcinoma cell lines, established through exposure to increasing concentrations of 5-FU, showed upregulation of Id1, IGF2, and E2F1. We hypothesized that these genes may play an important role in cancer chemoresistance. EXPERIMENTAL DESIGN In vitro and in vivo functional assays were performed to study the effects of Id1-E2F1-IGF2 signaling in chemoresistance. Quantitative real-time PCR, Western blotting, immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter assays were used to investigate the molecular mechanisms by which Id1 regulates E2F1 and by which E2F1 regulates IGF2. Clinical specimens, tumor tissue microarray, and Gene Expression Omnibus datasets were used to analyze the correlations between gene expressions and the relationships between expression profiles and patient survival outcomes. RESULTS Id1 conferred 5-FU chemoresistance through E2F1-dependent induction of thymidylate synthase expression in esophageal cancer cells and tumor xenografts. Mechanistically, Id1 protects E2F1 protein from degradation and increases its expression by binding competitively to Cdc20, whereas E2F1 mediates Id1-induced upregulation of IGF2 by binding directly to the IGF2 promoter and activating its transcription. The expression level of E2F1 was positively correlated with that of Id1 and IGF2 in human cancers. More importantly, concurrent high expression of Id1 and IGF2 was associated with unfavorable patient survival in multiple cancer types. CONCLUSIONS Our findings define an intricate E2F1-dependent mechanism by which Id1 increases thymidylate synthase and IGF2 expressions to promote cancer chemoresistance. The Id1-E2F1-IGF2 regulatory axis has important implications for cancer prognosis and treatment.
Collapse
Affiliation(s)
- Bin Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China. Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wen Wen Xu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Xin Yuan Guan
- Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yan Ru Qin
- Department of Clinical Oncology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Simon Law
- Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. Department of Surgery The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Nikki Pui Yue Lee
- Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. Department of Surgery The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kin Tak Chan
- Department of Surgery The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Pui Ying Tam
- Department of Surgery The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuk Yin Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Kwok Wah Chan
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China. Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hiu Fung Yuen
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Qing Yu He
- Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| | - Annie L M Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China. Centre for Cancer Research, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
40
|
Zhang X, Xu L, Yin L, Qi Y, Xu Y, Han X, Peng J. Quantitative chemical proteomics for investigating the biomarkers of dioscin against liver fibrosis caused by CCl4 in rats. Chem Commun (Camb) 2015; 51:11064-11067. [PMID: 26069897 DOI: 10.1039/c4cc09160d] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
In the present work, the effect of dioscin against liver fibrosis in rats caused by carbon tetrachloride (CCl4) was confirmed. Then, the differentially expressed proteins from rat liver were identified using two-dimensional differential in-gel electrophoresis technology. Ten new biomarkers including protein disulfide isomerase A3, selenium-binding protein 1, glutamine synthetase, senescence marker protein 30, hemopexin, keratin 8, keratin 18, vimentin, Annexin A5 and dermatopontin associated with liver fibrosis were found and validated, and new insights through affecting multiple drug targets and biological processes were also provided to reveal the mechanisms of dioscin against hepatic fibrosis for the first time.
Collapse
Affiliation(s)
- Xiaoling Zhang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| | | | | | | | | | | | | |
Collapse
|
41
|
Chigaev A. Does aberrant membrane transport contribute to poor outcome in adult acute myeloid leukemia? Front Pharmacol 2015; 6:134. [PMID: 26191006 PMCID: PMC4489100 DOI: 10.3389/fphar.2015.00134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/15/2015] [Indexed: 12/31/2022] Open
Abstract
Acute myeloid leukemia in adults is a highly heterogeneous disease. Gene expression profiling performed using unsupervised algorithms can be used to distinguish specific groups of patients within a large patient cohort. The identified gene expression signatures can offer insights into underlying physiological mechanisms of disease pathogenesis. Here, the analysis of several related gene expression clusters associated with poor outcome, worst overall survival and highest rates of resistant disease and obtained from the patients at the time of diagnosis or from previously untreated individuals is presented. Surprisingly, these gene clusters appear to be enriched for genes corresponding to proteins involved in transport across membranes (transporters, carriers and channels). Several ideas describing the possible relationship of membrane transport activity and leukemic cell biology, including the "Warburg effect," the specific role of chloride ion transport, direct "import" of metabolic energy through uptake of creatine phosphate, and modification of the bone marrow niche microenvironment are discussed.
Collapse
Affiliation(s)
- Alexandre Chigaev
- Department of Pathology and Cancer Center, University of New Mexico Health Sciences Center, University of New Mexico Albuquerque, NM, USA
| |
Collapse
|
42
|
The subcellular location of selenoproteins and the impact on their function. Nutrients 2015; 7:3938-48. [PMID: 26007340 PMCID: PMC4446787 DOI: 10.3390/nu7053938] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 01/31/2023] Open
Abstract
Most human selenium containing proteins contain selenium in the form of the amino acid selenocysteine, which is encoded in the corresponding mRNA as a UGA codon. Only a few non-selenocysteine containing selenoproteins are present and the nature of the association with selenium is not well understood. This review focuses on two selenocysteine-containing proteins that are members of the glutathione peroxidase family, GPx-1 and GPx-4, and the selenium-associated protein referred to as Selenium Binding Protein 1. Each of these proteins have been described to reside in two or more cellular compartments, and in the case of GPx-1 and SBP1, interact with each other. The enzymatic activity of GPx-1 and GPx-4 have been well described, but it is less clear how their cellular location impacts the health related phenotypes associated with activities, while no catalytic function is assigned to SBP1. The distribution of these proteins is presented as is the possible consequences of that compartmentalization.
Collapse
|
43
|
Quantitative proteomic analysis reveals that anti-cancer effects of selenium-binding protein 1 in vivo are associated with metabolic pathways. PLoS One 2015; 10:e0126285. [PMID: 25974208 PMCID: PMC4431778 DOI: 10.1371/journal.pone.0126285] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/31/2015] [Indexed: 12/03/2022] Open
Abstract
Previous studies have shown the tumor-suppressive role of selenium-binding protein 1 (SBP1), but the underlying mechanisms are unclear. In this study, we found that induction of SBP1 showed significant inhibition of colorectal cancer cell growth and metastasis in mice. We further employed isobaric tags for relative and absolute quantitation (iTRAQ) to identify proteins that were involved in SBP1-mediated anti-cancer effects in tumor tissues. We identified 132 differentially expressed proteins, among them, 53 proteins were upregulated and 79 proteins were downregulated. Importantly, many of the differentially altered proteins were associated with lipid/glucose metabolism, which were also linked to Glycolysis, MAPK, Wnt, NF-kB, NOTCH and epithelial-mesenchymal transition (EMT) signaling pathways. These results have revealed a novel mechanism that SBP1-mediated cancer inhibition is through altering lipid/glucose metabolic signaling pathways.
Collapse
|
44
|
Jeong JY, Zhou JR, Gao C, Feldman L, Sytkowski AJ. Human selenium binding protein-1 (hSP56) is a negative regulator of HIF-1α and suppresses the malignant characteristics of prostate cancer cells. BMB Rep 2015; 47:411-6. [PMID: 24874852 PMCID: PMC4163856 DOI: 10.5483/bmbrep.2014.47.7.104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Indexed: 12/22/2022] Open
Abstract
In the present study, we demonstrate that ectopic expression of 56-kDa human selenium binding protein-1 (hSP56) in PC-3 cells that do not normally express hSP56 results in a marked inhibition of cell growth in vitro and in vivo. Down-regulation of hSP56 in LNCaP cells that normally express hSP56 results in enhanced anchorage-independent growth. PC-3 cells expressing hSP56 exhibit a significant reduction of hypoxia inducible protein (HIF)-1α protein levels under hypoxic conditions without altering HIF-1α mRNA (HIF1A) levels. Taken together, our findings strongly suggest that hSP56 plays a critical role in prostate cells by mechanisms including negative regulation of HIF-1α, thus identifying hSP56 as a candidate anti-oncogene product. [BMB Reports 2014; 47(7): 411-416]
Collapse
Affiliation(s)
- Jee-Yeong Jeong
- Laboratory for Cell and Molecular Biology, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Biochemistry and Cancer Research Institute, Kosin University College of Medicine, Busan 602-703, Korea
| | - Jin-Rong Zhou
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chong Gao
- Laboratory for Cell and Molecular Biology, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laurie Feldman
- Laboratory for Cell and Molecular Biology, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Arthur J Sytkowski
- Laboratory for Cell and Molecular Biology, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; Oncology Therapeutic Area, Quintiles Translational, Arlington, MA 02476,USA
| |
Collapse
|
45
|
Wang Y, Fang W, Huang Y, Hu F, Ying Q, Yang W, Xiong B. Reduction of selenium-binding protein 1 sensitizes cancer cells to selenite via elevating extracellular glutathione: a novel mechanism of cancer-specific cytotoxicity of selenite. Free Radic Biol Med 2015; 79:186-96. [PMID: 25445402 DOI: 10.1016/j.freeradbiomed.2014.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/14/2014] [Accepted: 11/20/2014] [Indexed: 01/14/2023]
Abstract
Selenium is an essential trace element and has been extensively studied for preventive effects on cancers. Recent emerging evidence has also shown that selenium at supranutritional dosage has a preferential cytotoxicity in cancer cells and chemotherapeutic drug-resistant cells, but the underlying mechanisms remain largely unknown. This study was to investigate the roles of two distinct representatives of selenium-containing proteins, selenium-binding protein 1 (SBP1) and glutathione peroxidase 1 (GPX1), in selenite-mediated cancer-specific cytotoxicity. We found that there was a significantly inverse correlation between SBP1 and GPX1 protein level in human breast cancers and adjacent matched nontumor tissues (Pearson r=-0.4347, P=0.0338). Ectopic expression of GPX1 enhanced selenite cytotoxicity through down-regulation of SBP1, and SBP1 was likely to be a crucial determinant for selenite-mediated cytotoxicity. Reduction of SBP1 in cancer cells and epirubicin-resistant cells on selenite exposure resulted in a dramatic increase in the generation of hydrogen peroxide and superoxide anion, which in turn caused oxidative stress and triggered apoptosis. Furthermore, knockdown SBP1 by small interfering RNA increased selenite sensitivity by elevating extracellular glutathione (GSH), which spontaneously reacted with selenite and led to the rapid depletion of selenium (IV) in growth medium and the high-affinity uptake of selenite. In conclusion, these findings would improve our understanding of the roles of selenium-containing proteins in selenite-mediated cytotoxicity, and revealed a potent mechanism of the selective cytotoxicity of selenite in cancer cells and drug-resistant cells, in which SBP1 was likely to play an important role in modulating the extracellular microenvironment by regulating the levels of extracellular GSH.
Collapse
Affiliation(s)
- Yulei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Ying Huang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Department of Oncology, the Fifth Hospital, Wuhan, Hubei, 430051, China
| | - Fen Hu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Qi Ying
- Department of Pathology, University of Illinois at Chicago, IL 60612, USA
| | - Wancai Yang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Department of Pathology, University of Illinois at Chicago, IL 60612, USA.
| | - Bin Xiong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China; Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, Hubei, 430071, China.
| |
Collapse
|
46
|
Abstract
INTRODUCTION Esophageal adenocarcinomas (EAC) are aggressive cancers that are increasing in incidence and associated with a poor prognosis. The identification of highly expressed genes in EAC relative to metaplastic Barrett's esophagus (BE) may provide new targets for novel early cancer detection strategies using endoscopically administered, fluorescently labeled peptides. METHODS Gene expression analysis of BE and EACs were used to identify the cell surface marker transglutaminase 2 (TGM2) as overexpressed in cancer. The expression of two major isoforms of TGM2 was determined by qRT-polymerase chain reaction in an independent cohort of 128 EACs. Protein expression was confirmed by tissue microarrays and immunoblot analysis of EAC cell lines. TGM2 DNA copy number was assessed using single nucleotide polymorphism microarrays and confirmed by qPCR. TGM2 expression in neoadjuvantly treated EACs and following small interfering RNA-mediated knockdown in cisplatin-treated EAC cells was used to determine its possible role in chemoresistance. RESULTS TGM2 is overexpressed in 15 EACs relative to 26 BE samples. Overexpression of both TGM2 isoforms was confirmed in 128 EACs and associated with higher tumor stage, poor differentiation, and increased inflammatory and desmoplastic response. Tissue microarrays and immunohistochemistry confirmed elevated TGM2 protein expression in EAC. Single nucleotide polymorphism and qPCR analysis revealed increased TGM2 gene copy number as one mechanism underlying elevated TGM2 expression. TGM2 was highly expressed in resistant EAC after patient treatment with neoadjuvant chemotherapy/radiation suggesting a role for TGM2 in chemoresistance. CONCLUSION TGM2 may be a useful cell surface biomarker for early detection of EAC.
Collapse
|
47
|
Modulators of cellular senescence: mechanisms, promises, and challenges from in vitro studies with dietary bioactive compounds. Nutr Res 2014; 34:1017-35. [DOI: 10.1016/j.nutres.2014.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 12/11/2022]
|
48
|
Schild F, Kieffer-Jaquinod S, Palencia A, Cobessi D, Sarret G, Zubieta C, Jourdain A, Dumas R, Forge V, Testemale D, Bourguignon J, Hugouvieux V. Biochemical and biophysical characterization of the selenium-binding and reducing site in Arabidopsis thaliana homologue to mammals selenium-binding protein 1. J Biol Chem 2014; 289:31765-31776. [PMID: 25274629 PMCID: PMC4231655 DOI: 10.1074/jbc.m114.571208] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 09/17/2014] [Indexed: 12/19/2022] Open
Abstract
The function of selenium-binding protein 1 (SBP1), present in almost all organisms, has not yet been established. In mammals, SBP1 is known to bind the essential element selenium but the binding site has not been identified. In addition, the SBP family has numerous potential metal-binding sites that may play a role in detoxification pathways in plants. In Arabidopsis thaliana, AtSBP1 over-expression increases tolerance to two toxic compounds for plants, selenium and cadmium, often found as soil pollutants. For a better understanding of AtSBP1 function in detoxification mechanisms, we investigated the chelating properties of the protein toward different ligands with a focus on selenium using biochemical and biophysical techniques. Thermal shift assays together with inductively coupled plasma mass spectrometry revealed that AtSBP1 binds selenium after incubation with selenite (SeO3(2-)) with a ligand to protein molar ratio of 1:1. Isothermal titration calorimetry confirmed the 1:1 stoichiometry and revealed an unexpectedly large value of binding enthalpy suggesting a covalent bond between selenium and AtSBP1. Titration of reduced Cys residues and comparative mass spectrometry on AtSBP1 and the purified selenium-AtSBP1 complex identified Cys(21) and Cys(22) as being responsible for the binding of one selenium. These results were validated by site-directed mutagenesis. Selenium K-edge x-ray absorption near edge spectroscopy performed on the selenium-AtSBP1 complex demonstrated that AtSBP1 reduced SeO3(2-) to form a R-S-Se(II)-S-R-type complex. The capacity of AtSBP1 to bind different metals and selenium is discussed with respect to the potential function of AtSBP1 in detoxification mechanisms and selenium metabolism.
Collapse
Affiliation(s)
- Florie Schild
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359
| | - Sylvie Kieffer-Jaquinod
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Biologie à Grande Echelle, Université Grenoble Alpes, CEA, INSERM, 17 rue des Martyrs, F-38000 Grenoble, France
| | - Andrés Palencia
- European Molecular Biology Laboratory Outstation, 71 avenue des Martyrs, F-38042 Grenoble, France and Unit for Virus Host-Cell Interactions, Université Grenoble Alpes-EMBL-CNRS, 71 avenue des Martyrs, 38042 France
| | - David Cobessi
- Université Grenoble Alpes, CEA, CNRS, Direction des Sciences du Vivant, Institut de Biologie Structurale, 6 rue Jules Horowitz, F-38044 Grenoble, France
| | - Géraldine Sarret
- Université Grenoble Alpes, CNRS & IRD, ISTerre, BP 53, F-38041 Grenoble, France
| | - Chloé Zubieta
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359
| | - Agnès Jourdain
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359
| | - Renaud Dumas
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359
| | - Vincent Forge
- Laboratoire de Chimie et Biologie des Métaux, Université Grenoble Alpes, CEA, CNRS, Institut de Recherches en Technologies et Sciences pour le Vivant, 17 rue des Martyrs, F-38000 Grenoble, France, and
| | - Denis Testemale
- Université Grenoble Alpes, CNRS, Institut NEEL, 25 rue des Martyrs, F-38042 Grenoble, France
| | - Jacques Bourguignon
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359
| | - Véronique Hugouvieux
- Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire de Physiologie Cellulaire et Végétale, CEA, Université Grenoble Alpes, CNRS UMR5168, INRA USC1359,.
| |
Collapse
|
49
|
Wang N, Chen Y, Yang X, Jiang Y. Selenium-binding protein 1 is associated with the degree of colorectal cancer differentiation and is regulated by histone modification. Oncol Rep 2014; 31:2506-14. [PMID: 24737289 DOI: 10.3892/or.2014.3141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/24/2014] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to examine the regulation of selenium binding protein 1 (SELENBP1) expression in colorectal cancer (CRC). Samples of cancer tissue and adjacent normal mucosa were collected from 83 CRC patients, and analyzed for SELENBP1 expression by 2D-DIGE, immunoblotting, RT-PCR and immunostaining. Expression levels of SELENBP1, carcinoembryonic antigen (CEA) and alkaline phosphatase (AKP) were determined in cultures of human colon cancer cell lines (SW480, SW620 and HT29) folllowing treatment with i) sodium butyrate (NaB, 2 mM), a differentiation inducer; ii) Trichostatin A (TSA, 0.3 µM), a histone deacetylase inhibitor; or iii) 5'-aza-2'-deoxycytidine (5-Aza-dC, 5 µM), a DNA methylation inhibitor. SELENBP1 expression was found to be downregulated (2.54-fold) in the CRC samples as determined by 2D-DIGE and confirmed by immunoblotting and RT-PCR. SELENBP1 expression was correlated with the degree of differentiation, but not with TNM stage or lymph node metastasis, and was higher in benign polyps (1.97±0.57) than in CRC tissues (0.96±0.59). In the CRC cell lines, NaB treatment led to the upregulation of SELENBP1, CEA and AKP when compared with the untreated cells (2.24- to 4.82-fold). SELENBP1 was also upregulated in cells treated with TSA alone (1.25- to 3.64-fold), or in combination with 5-Aza-dC (1.32- to 4.13-fold). In CRC, the downregulated SELENBP1 expression was reactivated by inducing differentiation. Therefore, SELENBP1 is a potential pharmacological target for individualized CRC treatment.
Collapse
Affiliation(s)
- Ning Wang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yang Chen
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinghua Yang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi Jiang
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
50
|
Liu Y, Tennant DA, Zhu Z, Heath JK, Yao X, He S. DiME: a scalable disease module identification algorithm with application to glioma progression. PLoS One 2014; 9:e86693. [PMID: 24523864 PMCID: PMC3921127 DOI: 10.1371/journal.pone.0086693] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/13/2013] [Indexed: 11/21/2022] Open
Abstract
Disease module is a group of molecular components that interact intensively in the disease specific biological network. Since the connectivity and activity of disease modules may shed light on the molecular mechanisms of pathogenesis and disease progression, their identification becomes one of the most important challenges in network medicine, an emerging paradigm to study complex human disease. This paper proposes a novel algorithm, DiME (Disease Module Extraction), to identify putative disease modules from biological networks. We have developed novel heuristics to optimise Community Extraction, a module criterion originally proposed for social network analysis, to extract topological core modules from biological networks as putative disease modules. In addition, we have incorporated a statistical significance measure, B-score, to evaluate the quality of extracted modules. As an application to complex diseases, we have employed DiME to investigate the molecular mechanisms that underpin the progression of glioma, the most common type of brain tumour. We have built low (grade II) - and high (GBM) - grade glioma co-expression networks from three independent datasets and then applied DiME to extract potential disease modules from both networks for comparison. Examination of the interconnectivity of the identified modules have revealed changes in topology and module activity (expression) between low- and high- grade tumours, which are characteristic of the major shifts in the constitution and physiology of tumour cells during glioma progression. Our results suggest that transcription factors E2F4, AR and ETS1 are potential key regulators in tumour progression. Our DiME compiled software, R/C++ source code, sample data and a tutorial are available at http://www.cs.bham.ac.uk/~szh/DiME.
Collapse
Affiliation(s)
- Yunpeng Liu
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
| | - Daniel A. Tennant
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zexuan Zhu
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - John K. Heath
- Centre for Systems Biology, School of Biological Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Xin Yao
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
| | - Shan He
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
- Centre for Systems Biology, School of Biological Sciences, University of Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|